Mechanisms of glioma formation: iterative perivascular glioma growth and invasion leads to tumor progression, VEGF-independent vascularization, and resistance to antiangiogenic therapy

Gregory J Baker, Viveka Nand Yadav, Sebastien Motsch, Carl Koschmann, Anda-Alexandra Calinescu, Yohei Mineharu, Sandra Ines Camelo-Piragua, Daniel Orringer, Serguei Bannykh, Wesley S Nichols, Ana C deCarvalho, Tom Mikkelsen, Maria G Castro, Pedro R Lowenstein, Gregory J Baker, Viveka Nand Yadav, Sebastien Motsch, Carl Koschmann, Anda-Alexandra Calinescu, Yohei Mineharu, Sandra Ines Camelo-Piragua, Daniel Orringer, Serguei Bannykh, Wesley S Nichols, Ana C deCarvalho, Tom Mikkelsen, Maria G Castro, Pedro R Lowenstein

Abstract

As glioma cells infiltrate the brain they become associated with various microanatomic brain structures such as blood vessels, white matter tracts, and brain parenchyma. How these distinct invasion patterns coordinate tumor growth and influence clinical outcomes remain poorly understood. We have investigated how perivascular growth affects glioma growth patterning and response to antiangiogenic therapy within the highly vascularized brain. Orthotopically implanted rodent and human glioma cells are shown to commonly invade and proliferate within brain perivascular space. This form of brain tumor growth and invasion is also shown to characterize de novo generated endogenous mouse brain tumors, biopsies of primary human glioblastoma (GBM), and peripheral cancer metastasis to the human brain. Perivascularly invading brain tumors become vascularized by normal brain microvessels as individual glioma cells use perivascular space as a conduit for tumor invasion. Agent-based computational modeling recapitulated biological perivascular glioma growth without the need for neoangiogenesis. We tested the requirement for neoangiogenesis in perivascular glioma by treating animals with angiogenesis inhibitors bevacizumab and DC101. These inhibitors induced the expected vessel normalization, yet failed to reduce tumor growth or improve survival of mice bearing orthotopic or endogenous gliomas while exacerbating brain tumor invasion. Our results provide compelling experimental evidence in support of the recently described failure of clinically used antiangiogenics to extend the overall survival of human GBM patients.

Copyright © 2014 Neoplasia Press, Inc. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
GL26-Cit glioma uses preexisting brain microvasculature as a scaffold for brain tumor invasion. (A) Tumor growth timeline indicating the time points analyzed over the first 120 hours of intracranial GL26-Cit glioma growth. Colored bars represent time frames corresponding to the three distinct phases of early intracranial GL26-Cit glioma growth. (B) Representative fluorescence scanning confocal micrographs of GL26-Cit glioma in the RAG1−/− mouse striatum imaged at the nine predetermined time points indicated in A. (C) Representative fluorescence scanning confocal micrographs of mouse brain microvessels from RA/EGxdelCre (left) and Rag1tm1MomTg(TIE2GFP)287Sato/J (i.e., Tie-2-GFP) (right) mice used to determine the fractal dimension of normal mouse brain microvasculature. Fractal dimension values (D values) for each micrograph are shown. (D) Average D values of gliomas from RAG1−/− mice at 0.25 and 48 hpi (n = 6 mice per group) compared to the average D value of brain microvasculature from the two GFP+ mouse strains represented in C (n = 8; four mice per strain with five distinct striatal regions imaged per mouse). ***P < .0001 versus tumors 0.25 hpi; one-way analysis of variance followed by Tukey post-test. Data represent the mean ± SEM. (E) Fluorescence scanning confocal micrograph of GL26-Cit glioma 48 hpi into the RA/EGxdelCre mouse striatum. GFP+ mouse brain microvessels have been pseudocolored red. White arrowheads point to several examples of microvasculature-associated glioma invasion. (F) Intravital multiphoton micrograph of GL26-Cit glioma imaged through an intracranial window 120 hpi into the somatosensory cortex of the RAG1−/− mouse brain (imaging depth = 129 μm). Mouse brain microvasculature was labeled intraluminally by tail-vein injection of rhodamine B isothiocyanate–conjugated dextran (mw = 70 kDa) before imaging. White arrowheads point to numerous examples of microvasculature-associated tumor invasion. (G) GL26-Cit after 58 days post-tumor implantation into the RAG1−/− mouse brain shown at low (left) and high (right) magnification. Kaplan-Meier survival analysis (lower right) demonstrates that initial injections of low numbers of glioma cells extend animal survival to study late-stage tumor invasion. (H) TEM at low power (top image) showing longitudinal and transverse capillary segments (pseudocolored red) enveloped by perivascularly invading GL26-Cit glioma cells (cytoplasm, green; nuclei, blue) 48 hpi in the RAG1−/− mouse brain. Tumor cells displace the immediately surrounding neuropil as they enter the perivascular space. White arrows indicate USPIOs used to label tumor cells in vitro before implantation. Higher magnification image of the area outlined by the white box is shown below. Black arrowheads in lower micrograph point to the vascular basement membrane covering the adluminal surface of the vascular endothelium. L, vessel lumen.
Figure 2
Figure 2
Perivascular invasion is an iterative growth process. (A) Scanning fluorescence confocal micrographs of GL26-Cit glioma cells from 0.25 to 120 hours in the RA/EGxdelCre mouse brain. Glioma cells are shown in green; brain microvessels are shown in red. (B) Schematic representation of the process of perivascular glioma growth and invasion beginning immediately post-tumor implantation (Step 1-2). Implanted glioma cells initially infiltrate throughout the perivascular space (Step 3-4). Cell division causes normal brain parenchyma present between adjacent microvessels to be displaced by neoplastic tissue, leaving normal brain microvessels within the tumor core (Step 4-5). Iterations of further perivascular invasion and cell division produce well-vascularized brain tumors throughout tumor growth (Steps 6-8).
Figure 3
Figure 3
Brain tumors of diverse species and cellular origin exhibit perivascular invasion. (A) Fluorescence scanning confocal micrographs of fluorescently modified CNS-1-Cit rat glioma cells in the brain of syngeneic Lewis rats after 48 hpi (left image) and 72 hpi (right two images). Brain microvasculature was immunolabeled with anti-laminin antibodies, a marker of the vascular basement membrane. White arrowheads point to distinct examples of microvasculature-associated tumor invasion. (B) Nissl staining of human U251 glioma cells in NU/J mice 72 hpi. Black boxes in each brightfield micrograph outline the field of view shown to the right at increasing magnification. Black arrows in the far right image point to tumor cell bodies heavily stained with Nissl that assume vascular morphology as they migrate away from the main tumor mass. N, tumor necrosis. (C) Fluorescence scanning confocal micrographs of HF2303 primary human GBM cancer stem cells 14 and 143 dpi into the RAG1−/− mouse striatum. Primary human GBM cancer stem cells are immunoreactive for the neural stem cell marker nestin. Brain microvessels have been labeled with anti-CD31 antibodies. White arrowheads indicate several examples of microvasculature-associated tumor invasion. T, bulk tumor mass. (D) Tissue biopsy from a human patient bearing mammary carcinoma metastasis to the brain. The metastatic lesion (purple) seen at low magnification (top left panel) can be seen adjacent to a large superficial blood vessel heavily infiltrated with perivascular tumor cells (white arrowheads). Higher magnification (bottom left panel) of the area outlined by the black box in the panel above reveals the invasive margin of the metastatic lesion. Largest panel to the right shows the invasive margin outlined by the black box in the bottom left image at × 20 magnification. White arrowheads point to several examples of invasive cancer cells (purple) surrounding brain microvessels (seen in red due to the presence of intraluminal red blood cells).
Figure 4
Figure 4
De novo mouse GBM and clinical GBM biopsies exhibit perivascular invasion. (A) De novo mouse GBM generated using the Sleeping Beauty transposase system at 60 days post-plasmid injection. At the center, a 5 × mosaic epifluorescence micrograph of a coronal mouse brain tissue section is shown. Tumor cells have been labeled using anti-nestin antibodies. Brain microvasculature has been labeled with anti–Von Willebrand factor antibodies. 4',-diamidino-2-phenylindole (DAPI) has been used as a counterstain (blue). Numbered white boxes within the central epifluorescence micrograph correspond to respectively numbered higher magnification fluorescence scanning confocal micrographs surrounding it. Several examples of perivascular tissue invasion are indicated by white arrowheads. (B) Human GBM clinical biopsy. A 5 × mosaic epifluorescence micrograph of a tissue biopsy immunolabeled with anti-vimentin antibodies, anti–Von Willebrand factor antibodies, and DAPI (blue) as a counterstain is shown. The zone of transition between tumor tissue (left) and normal brain (right) lies within the white dashed lines overlying each fluorescence channel. Panels below the high-magnification fluorescence scanning confocal micrographs correspond to the dashed white boxes from the epifluorescence images above. White arrowheads in each confocal micrograph point to examples of perivascular tissue invasion.
Figure 5
Figure 5
In silico computational modeling mimics perivascular brain tumor growth. (A) Schematic representation of an agent-based computational model describing perivascular brain tumor growth and invasion. Individual glioma cells move toward nearby blood vessels under the influence of a velocity field vPω→iT∇gx→i that mimics the predilection of glioma cells for blood vessels (an underlying assumption of the model). Cells move by a correlated random walk with constant speed toward nearby blood vessels. Once tumor cells make contact with a vessel, they slow their migration speed and proliferate at a rate μb. Tumor cells not associated with a blood vessel eventually undergo cell death at a rate μd and have an average life expectancy of 1/μd. BV, blood vessel. (B) A table summarizing the parameters used in the agent-based model. The respective symbols and optimal values are indicated for each parameter. (C) Representative snapshots of simulated brain tumors corresponding to the three phases of early perivascular tumor growth. The microvasculature domain (vessels only) has been taken from a fluorescence scanning confocal micrograph of RA/EGxdelCre mouse brain microvessels. The simulated tumor initially consists of 1 × 103 cells centered with the microvasculature domain at t = 0, simulating preinvasive phase I glioma soon after implantation (top right panel). Tumor cells then begin to migrate toward nearby microvessels resulting in a strong correlation between the density of tumor cells and blood vessels, causing the characteristic branch-like morphology of phase II glioma (bottom left panel). Tumor cells making contact with a blood vessel rapidly proliferate and fill the gaps between adjacent blood vessels as the total number of cells grows exponentially. As a result, the tumor center widens and becomes hypercellular while maintaining extensive microvascular contact at the tumor borders in a fashion that mimics phase III glioma (bottom right panel).
Figure 6
Figure 6
Perivascular glioma invasion obviates the requirement for tumor neoangiogenesis to support continual intracranial growth. (A) Representative fluorescence scanning confocal micrographs of GL26-Cit glioma over the initial 144 hpi in the RAG1−/− mouse brain. Mice were treated with non-specific control IgG (top row) or the VEGF-A blocking antibody bevacizumab (bottom row). (B and C) Quantification of time point–matched tumors (n = 30; three tumors per group per time point) reveals no significant difference in overall tumor size as measured by the average tumor area in pixels (B) or morphology as measured by tumor fractal dimension (C) between control IgG– and bevacizumab-treated mice over the 144-hour analysis period. The P value between treatment groups was > .05 at each time point analyzed by two-way analysis of variance followed by Bonferroni post-tests. (D) Immunohistochemistry on GL26-Cit bearing mouse brain tissue sections labeled with the vascular markers CD31 and laminin (cyan) after 144 hours of treatment with control IgG (left) or bevacizumab (right). Solid white tumor outlines in the CD31 channels circumscribe tumor-associated microvasculature and reveal the vessel “normalization” effect in the bevacizumab treatment group. Dashed white boxes outline the area imaged at higher magnification in the two images below each treatment group, further revealing the anatomic detail of vessel preservation in the bevacizumab-treated group. (E) Kaplan-Meier survival analysis of RAG1−/− mice bearing GL26-Cit glioma treated with bevacizumab (n = 5) or control IgG (n = 5). Mantel-Cox log-rank test detected no significant survival difference between the bevacizumab and control IgG treatment groups (P = .3560). (F) Kaplan-Meier survival analysis of RAG1−/− mice bearing endogenous brain tumors generated de novo using the Sleeping Beauty transposase system treated with bevacizumab (n = 9) or control IgG (n = 8). Mantel-Cox log-rank test detected no significant survival difference between the bevacizumab and control IgG treatment groups (P = .5240).
Figure 7
Figure 7
VEGFR-2 blockade with DC101 fails to inhibit tumor growth over 144 hours in vivo or improve survival alone, or as adjuvant therapy. (A) Representative fluorescence scanning confocal micrographs of GL26-Cit tumors in RAG1−/− mice treated with control IgG (top row) or DC101 (bottom row) at a dose of 40 mg/kg delivered i.p. twice weekly over 144 hours (n = 30; three mice per time point per treatment group). No discernable difference in tumor size or morphology is noted at each time point analyzed. (B) Kaplan-Meier survival analysis comparing C57BL/6J mice bearing GL26 gliomas treated with DC101 or control IgG administered three times (red arrows) before (B), or after (C), treatment with Ad-TK/Ad-Flt3L adenovirally mediated cytotoxic gene therapy. Saline treatment alone was used as a negative control. Mantel-Cox log-rank test detected no significant survival difference between Ad-TK/Ad-Flt3L plus DC101 and Ad-TK/Ad-Flt3L plus control IgG treatment either before (P = .8124) or after (P = .5123) treatment with Ad-TK/Ad-Flt3L.
Figure 8
Figure 8
Bevacizumab does not significantly improve the survival of RAG1−/− mice bearing HF2303 primary human GBM stem cells. (A) Kaplan-Meier survival analysis of RAG1−/− mice bearing human HF2303 GBM stem cells treated with bevacizumab or non-specific control IgG. Mantel-Cox log-rank test detected no significant survival difference between the bevacizumab and control IgG treatment groups (P = .0995). Each tick denotes 1 of the 54 total doses of bevacizumab administered throughout the study. n.s., not significant. (B) Representative photographs of gross brain morphology from moribund RAG1−/− mice treated with bevacizumab (left) or control IgG (right) both before (top images) and after (bottom images) sectioning. Note the extensive microvascular hemorrhage associated with tumor neoangiogenesis in the control IgG treatment group, which is completely absent in bevacizumab-treated brain tumors. (C) 5 × mosaic epifluorescence micrographs of coronal brain tissue sections from the mouse brains shown in B. Sections have been immunolabeled with human-specific anti-nestin antibodies to label tumor cells, anti-CD31 antibodies to label brain microvasculature, and DAPI as a counterstain to label nuclei (blue). Note that the contralateral striatum in the control IgG treatment group (denoted as St.; dashed white outlines) appears compressed, presumably due to nodular tumor growth while bevacizumab treatment induces massive invasion into the contralateral hemisphere. Dashed white boxes in the respective images correspond to high-magnification fluorescence scanning confocal micrographs shown below. Normalized tumor vascular is specifically seen in bevacizumab-treated human HF2303 tumors, while control IgG–treated brain tumors are characterized by extensive microvessel fragmentation.
Figure 9
Figure 9
Quantification of brain microvascular network density. (A) Microvessels within the C57BL/6J tumor-naïve striatum were immunolabeled with anti-CD31, anti-laminin (cyan), and anti–α-SMA vascular markers and display a calculated average intervessel distance (AIVDcalc) of 56.95 ± 17.2 μm. (B) Microvessels within the RA/EGxdelCre tumor-naïve striatum showing an AIVDcalc of 56.29 ± 14.7 μm, in good agreement with the values obtained in A, indicating a low degree of variability in this parameter between different mouse strains. (C) Analysis of invasive GL26-Cit glioma cells at various time points at the infiltrative tumor margin within the RA/EGxdelCre mouse striatum (microvessels shown in red). Composite images (top row) show close vascular contacts made by individual tumor cells at each time point analyzed (white arrows). Individual fluorescence channels (middle and bottom rows) show AIVDcalc and calculated average tumor cell diameters (ACDcalc) for the respective micrographs. Overall averages for AIVDcalc and ACDcalc were determined to be 49.99 and 21.45 μm, respectively. (D) Low-magnification fluorescence scanning confocal micrograph of striatal microvasculature in the tumor-naïve RA/EGxdelCre brain. The blue circle illustrates the large number of brain microvessels present within a sphere of 1 mm3, a volume below which many solid tumors are thought togrow avascularly (here, we assume that microvessel density does not change significantly ± 1 mm in the Z direction). The radius of the circle has been calculated by solving for r=3mm3/4π3 in the equation for the volume of a sphere measuring 1 mm3 (upper right corner). The white arrow indicates the radius (r = 0.6205 mm) of the 1-mm3 sphere. The combination of high microvessel density and perivascular tumor invasion is predicted to preclude an avascular phase of brain tumor growth.
Figure 10
Figure 10
Brain tumor autovascularization. (A) Schematic representation of a coronally sectioned mouse brain revealing perivascular glioma growth and invasion within the striatum. Individual glioma cells are shown in green with blue nuclei. The tumor mass is vascularized with preexisting brain microvessels due to invasive glioma cells entering perivascular channels at the tumor border. Brain microvasculature is classified as arterial or venule (blue). Arterials are further characterized by the presence of smooth muscle (green bands) around larger diameter vessels, while the venous system is covered by patchwork pericytes (green amoeboid cells) as observed empirically using vascular-specific antibodies in situ (see Figure S7). No predilection for invasive glioma cells to migrate in association with arterials versus venules is observed. (B and C) Detailed views of invasive (B) and central (C) portions of perivascular brain tumors. White boxes shown in A correspond to the respective regions in B and C. (B) Illustration of brain perivascular potential space being infiltrated by invasive glioma cells. Adjacent microvessels have an average intervessel distance (AIVD) of ~ 50 μm (red scale bar), while the average tumor cell diameter (ACD) measures ~ 20 μm (green scale bar) as calculated empirically. Dashed white arrows represent the small intervening distance separating adjacent microvessels at the level of the capillary plexus. (C) Illustration of glioma cell division within the perivascular space. Glioma cell division within the perivascular space causes the displacement of normal brain tissue (i.e., neuropil, NP) between adjacent microvessels. The intervening space is then replaced by tumor cells while preexisting brain microvessels simultaneously become trapped within the growing tumor mass. Iterations of tumor cell invasion and division within the perivascular space causes further engulfment of preexisting brain microvessels, leading well-vascularized tumors early in tumor development through a neoangiogenesis-independent process we refer to as “autovascularization”. (D) Details of the microanatomic arrangement at the tumor-vessel interface based on empirical observations.

References

    1. Dolecek TA, Propp JM, Stroup NE, Kruchko C. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2005–2009. Neuro Oncol. 2012;14(Suppl 5):v1–v49.
    1. Grossman SA, Ye X, Piantadosi S, Desideri S, Nabors LB, Rosenfeld M, Fisher J. Survival of patients with newly diagnosed glioblastoma treated with radiation and temozolomide in research studies in the United States. Clin Cancer Res. 2010;16:2443–2449.
    1. Scherer HJ. The forms of growth in gliomas and their practical significance. Brain. 1940;63:1–35.
    1. Scherer HJ. Structural development in gliomas. Am J Cancer. 1938;34:333–351.
    1. Winkler F, Kienast Y, Fuhrmann M, Von Baumgarten L, Burgold S, Mitteregger G, Kretzschmar H, Herms J. Imaging glioma cell invasion in vivo reveals mechanisms of dissemination and peritumoral angiogenesis. Glia. 2009;57:1306–1315.
    1. Farin A, Suzuki SO, Weiker M, Goldman JE, Bruce JN, Canoll P. Transplanted glioma cells migrate and proliferate on host brain vasculature: a dynamic analysis. Glia. 2006;53:799–808.
    1. Holash J, Maisonpierre PC, Compton D, Boland P, Alexander CR, Zagzag D, Yancopoulos GD, Wiegand SJ. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science. 1999;284:1994–1998.
    1. Du R, Lu KV, Petritsch C, Liu P, Ganss R, Passegue E, Song H, Vandenberg S, Johnson RS, Werb Z. HIF1α induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell. 2008;13:206–220.
    1. Blouw B, Song H, Tihan T, Bosze J, Ferrara N, Gerber HP, Johnson RS, Bergers G. The hypoxic response of tumors is dependent on their microenvironment. Cancer Cell. 2003;4:133–146.
    1. Rubenstein JL, Kim J, Ozawa T, Zhang M, Westphal M, Deen DF, Shuman MA. Anti-VEGF antibody treatment of glioblastoma prolongs survival but results in increased vascular cooption. Neoplasia. 2000;2:306–314.
    1. Kunkel P, Ulbricht U, Bohlen P, Brockmann MA, Fillbrandt R, Stavrou D, Westphal M, Lamszus K. Inhibition of glioma angiogenesis and growth in vivo by systemic treatment with a monoclonal antibody against vascular endothelial growth factor receptor-2. Cancer Res. 2001;61:6624–6628.
    1. de Groot JF, Fuller G, Kumar AJ, Piao Y, Eterovic K, Ji Y, Conrad CA. Tumor invasion after treatment of glioblastoma with bevacizumab: radiographic and pathologic correlation in humans and mice. Neuro Oncol. 2010;12:233–242.
    1. Carbonell WS, DeLay M, Jahangiri A, Park CC, Aghi MK. β1 integrin targeting potentiates antiangiogenic therapy and inhibits the growth of bevacizumab-resistant glioblastoma. Cancer Res. 2013;73:3145–3154.
    1. Clark AJ, Lamborn KR, Butowski NA, Chang SM, Prados MD, Clarke JL, McDermott MW, Parsa AT, Berger MS, Aghi MK. Neurosurgical management and prognosis of patients with glioblastoma that progresses during bevacizumab treatment. Neurosurgery. 2012;70:361–370.
    1. Wiesner SM, Decker SA, Larson JD, Ericson K, Forster C, Gallardo JL, Long C, Demorest ZL, Zamora EA, Low WC. De novo induction of genetically engineered brain tumors in mice using plasmid DNA. Cancer Res. 2009;69:431–439.
    1. Shaner NC, Steinbach PA, Tsien RY. A guide to choosing fluorescent proteins. Nat Methods. 2005;2:905–909.
    1. Constien R, Forde A, Liliensiek B, Grone HJ, Nawroth P, Hämmerling G, Arnold B. Characterization of a novel EGFP reporter mouse to monitor Cre recombination as demonstrated by a Tie2 Cre mouse line. Genesis. 2001;30:36–44.
    1. Motoike T, Loughna S, Perens E, Roman BL, Liao W, Chau TC, Richardson CD, Kawate T, Kuno J, Weinstein BM. Universal GFP reporter for the study of vascular development. Genesis. 2000;28:75–81.
    1. Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, Benveniste H, Vates GE, Deane R, Goldman SA. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid β. Sci Transl Med. 2012;4:147ra111.
    1. Zhang ET, Richards HK, Kida S, Weller RO. Directional and compartmentalised drainage of interstitial fluid and cerebrospinal fluid from the rat brain. Acta Neuropathol. 1992;83:233–239.
    1. Pollock H, Hutchings M, Weller RO, Zhang ET. Perivascular spaces in the basal ganglia of the human brain: their relationship to lacunes. J Anat. 1997;191(Pt 3):337–346.
    1. Flanagan SP. ‘Nude’, a new hairless gene with pleiotropic effects in the mouse. Genet Res. 1966;8:295–309.
    1. Binda E, Visioli A, Giani F, Lamorte G, Copetti M, Pitter KL, Huse JT, Cajola L, Zanetti N, DiMeco F. The EphA2 receptor drives self-renewal and tumorigenicity in stem-like tumor-propagating cells from human glioblastomas. Cancer Cell. 2012;22:765–780.
    1. deCarvalho AC, Nelson K, Lemke N, Lehman NL, Arbab AS, Kalkanis S, Mikkelsen T. Gliosarcoma stem cells undergo glial and mesenchymal differentiation in vivo. Stem Cells. 2010;28:181–190.
    1. Carbonell WS, Ansorge O, Sibson N, Muschel R. The vascular basement membrane as “soil” in brain metastasis. PLoS One. 2009;4:e5857.
    1. Fidler IJ, Yano S, Zhang RD, Fujimaki T, Bucana CD. The seed and soil hypothesis: vascularisation and brain metastases. Lancet Oncol. 2002;3:53–57.
    1. Camazine S. Princeton University Press; Princeton, NJ: 2001. Self-Organization in Biological Systems.
    1. Renshaw E, Henderson R. The correlated random-walk. J Appl Probab. 1981;18:403–414.
    1. Kloeden PE, Platen E. Springer-Verlag; Berlin; New York: 1992. Numerical Solution of Stochastic Differential Equations.
    1. Ferrara N, Hillan KJ, Gerber HP, Novotny W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat Rev Drug Discov. 2004;3:391–400.
    1. Kienast Y, von Baumgarten L, Fuhrmann M, Klinkert WE, Goldbrunner R, Herms J, Winkler F. Real-time imaging reveals the single steps of brain metastasis formation. Nat Med. 2010;16:116–122.
    1. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science. 2005;307:58–62.
    1. Jain RK, di Tomaso E, Duda DG, Loeffler JS, Sorensen AG, Batchelor TT. Angiogenesis in brain tumours. Nat Rev Neurosci. 2007;8:610–622.
    1. Batchelor TT, Sorensen AG, di Tomaso E, Zhang WT, Duda DG, Cohen KS, Kozak KR, Cahill DP, Chen PJ, Zhu M. AZD2171, a pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11:83–95.
    1. Witte L, Hicklin DJ, Zhu Z, Pytowski B, Kotanides H, Rockwell P, Böhlen P. Monoclonal antibodies targeting the VEGF receptor-2 (Flk1/KDR) as an anti-angiogenic therapeutic strategy. Cancer Metastasis Rev. 1998;17:155–161.
    1. Jiang F, Zhang X, Kalkanis SN, Zhang Z, Yang H, Katakowski M, Hong X, Zheng X, Zhu Z, Chopp M. Combination therapy with antiangiogenic treatment and photodynamic therapy for the nude mouse bearing U87 glioblastoma. Photochem Photobiol. 2008;84:128–137.
    1. Martens T, Laabs Y, Günther HS, Kemming D, Zhu Z, Witte L, Hagel C, Westphal M, Lamszus K. Inhibition of glioblastoma growth in a highly invasive nude mouse model can be achieved by targeting epidermal growth factor receptor but not vascular endothelial growth factor receptor-2. Clin Cancer Res. 2008;14:5447–5458.
    1. King GD, Muhammad AK, Curtin JF, Barcia C, Puntel M, Liu C, Honig SB, Candolfi M, Mondkar S, Lowenstein PR. Flt3L and TK gene therapy eradicate multifocal glioma in a syngeneic glioblastoma model. Neuro Oncol. 2008;10:19–31.
    1. King GD, Muhammad AG, Larocque D, Kelson KR, Xiong W, Liu C, Sanderson NS, Kroeger KM, Castro MG, Lowenstein PR. Combined Flt3L/TK gene therapy induces immunological surveillance which mediates an immune response against a surrogate brain tumor neoantigen. Mol Ther. 2011;19:1793–1801.
    1. Yu L, Wu X, Cheng Z, Lee CV, LeCouter J, Campa C, Fuh G, Lowman H, Ferrara N. Interaction between bevacizumab and murine VEGF-A: a reassessment. Invest Ophthalmol Vis Sci. 2008;49:522–527.
    1. Bock F, Onderka J, Dietrich T, Bachmann B, Kruse FE, Paschke M, Zahn G, Cursiefen C. Bevacizumab as a potent inhibitor of inflammatory corneal angiogenesis and lymphangiogenesis. Invest Ophthalmol Vis Sci. 2007;48:2545–2552.
    1. Hua J, Spee C, Kase S, Rennel ES, Magnussen AL, Qiu Y, Varey A, Dhayade S, Churchill AJ, Harper SJ. Recombinant human VEGF165b inhibits experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2010;51:4282–4288.
    1. Avisar I, Weinberger D, Kremer I. Effect of subconjunctival and intraocular bevacizumab injections on corneal neovascularization in a mouse model. Curr Eye Res. 2010;35:108–115.
    1. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med. 1971;285:1182–1186.
    1. Folkman J, Hochberg M. Self-regulation of growth in three dimensions. J Exp Med. 1973;138:745–753.
    1. Eberhard A, Kahlert S, Goede V, Hemmerlein B, Plate KH, Augustin HG. Heterogeneity of angiogenesis and blood vessel maturation in human tumors: implications for antiangiogenic tumor therapies. Cancer Res. 2000;60:1388–1393.
    1. Warburg O. On the origin of cancer cells. Science. 1956;123:309–314.
    1. Flavahan WA, Wu Q, Hitomi M, Rahim N, Kim Y, Sloan AE, Weil RJ, Nakano I, Sarkaria JN, Stringer BW. Brain tumor initiating cells adapt to restricted nutrition through preferential glucose uptake. Nat Neurosci. 2013;16:1373–1382.
    1. Wolfenson H, Lavelin I, Geiger B. Dynamic regulation of the structure and functions of integrin adhesions. Dev Cell. 2013;24:447–458.
    1. Presta LG, Chen H, O'Connor SJ, Chisholm V, Meng YG, Krummen L, Winkler M, Ferrara N. Humanization of an anti-vascular endothelial growth factor monoclonal antibody for the therapy of solid tumors and other disorders. Cancer Res. 1997;57:4593–4599.
    1. Wolburg H, Noell S, Fallier-Becker P, Mack AF, Wolburg-Buchholz K. The disturbed blood-brain barrier in human glioblastoma. Mol Aspects Med. 2012;33:579–589.
    1. Huveldt D, Lewis-Tuffin LJ, Carlson BL, Schroeder MA, Rodriguez F, Giannini C, Galanis E, Sarkaria JN, Anastasiadis PZ. Targeting Src family kinases inhibits bevacizumab-induced glioma cell invasion. PLoS One. 2013;8:e56505.
    1. Gilbert MR, Dignam JJ, Armstrong TS, Wefel JS, Blumenthal DT, Vogelbaum MA, Colman H, Chakravarti A, Pugh S, Won M. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N Engl J Med. 2014;370:699–708.
    1. Chinot OL, Wick W, Mason W, Henriksson R, Saran F, Nishikawa R, Carpentier AF, Hoang-Xuan K, Kavan P, Cernea D. Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma. N Engl J Med. 2014;370:709–722.
    1. Lucio-Eterovic AK, Piao Y, de Groot JF. Mediators of glioblastoma resistance and invasion during antivascular endothelial growth factor therapy. Clin Cancer Res. 2009;15:4589–4599.
    1. Candolfi M, Curtin JF, Nichols WS, Muhammad AG, King GD, Pluhar GE, McNiel EA, Ohlfest JR, Freese AB, Moore PF. Intracranial glioblastoma models in preclinical neuro-oncology: neuropathological characterization and tumor progression. J Neurooncol. 2007;85:133–148.

Source: PubMed

3
Abonnieren